Hostname: page-component-848d4c4894-pjpqr Total loading time: 0 Render date: 2024-07-01T01:59:11.242Z Has data issue: false hasContentIssue false

Nanotexturization of Ti-based implants in simulated body fluid: Influence of synthesis parameters on coating properties and kinetics of drug release

Published online by Cambridge University Press:  30 August 2019

Carlise Hannel Ferreira
Affiliation:
Department of Chemistry, Federal Technological University of Paraná (UTFPR), Pato Branco, PR 85503-390, Brazil
Anna Paulla Simon
Affiliation:
Department of Chemistry, Federal Technological University of Paraná (UTFPR), Pato Branco, PR 85503-390, Brazil
Vidiany Aparecida Queiroz Santos
Affiliation:
Department of Chemistry, Federal Technological University of Paraná (UTFPR), Pato Branco, PR 85503-390, Brazil
Andressa Rodrigues
Affiliation:
Department of Physics, Chemistry and Mathematics, UFSCar, Sorocaba, SP 18052-780, Brazil
Janaina Soares Santos
Affiliation:
Department of Physics, Chemistry and Mathematics, UFSCar, Sorocaba, SP 18052-780, Brazil
Francisco Trivinho-Strixino
Affiliation:
Department of Physics, Chemistry and Mathematics, UFSCar, Sorocaba, SP 18052-780, Brazil
Patrícia Teixeira Marques
Affiliation:
Department of Chemistry, Federal Technological University of Paraná (UTFPR), Pato Branco, PR 85503-390, Brazil
Mariana de Souza Sikora*
Affiliation:
Department of Chemistry, Federal Technological University of Paraná (UTFPR), Pato Branco, PR 85503-390, Brazil
*
a)Address all correspondence to this author. e-mail: [email protected]
Get access

Abstract

In the present study, TiO2NT coatings grown on simulated body fluid-based electrolyte were investigated as drug delivery devices. Nanotubes (NTs) were grown over commercially pure Ti and Ti6Al4V alloy. Morphology analysis showed that NTs in alloy samples present an inner diameter of 10 nm smaller in average than NTs grown over pure Ti. The surface wettability in water decreased with the anodizing time for both substrates. The application of coatings as drug delivery devices has been studied through the incorporation of ciprofloxacin. To control the drug release, collagen was used as the diffusional barrier. It was observed the drug release follows a Fick’s kinetics. Bioactivity assays showed the absence of hemolytic activity. The concentration of the drug during the release interval remained below the toxic concentration limit, presenting a bacteriostatic activity. All coatings prepared presented a high antibacterial activity, being the area of inhibition of bacterial growth above 13 times the area of the implant.

Type
Article
Copyright
Copyright © Materials Research Society 2019 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Gurtner, G.C., Callaghan, M.J., and Longaker, M.T.: Progress and potential for regenerative medicine. Annu. Rev. Med. 58, 299 (2007).CrossRefGoogle ScholarPubMed
Habazaki, H., Onodera, T., Fushimi, K., Konno, H., and Toyotake, K.: Spark anodizing of β-Ti alloy for wear-resistant coating. Surf. Coat. Technol. 201, 8730 (2007).CrossRefGoogle Scholar
Holzapfel, B.M., Reichert, J., Schantz, J., Gbureck, U., Rackwitz, L., Nöth, U., Jakob, F., Rudert, M., Groll, J., and Werner, D.: How smart do biomaterials need to be? A translational science and clinical point of view. Adv. Drug Delivery Rev. 65, 581–603 (2012).Google Scholar
Murugan, R. and Ramakrishna, S.: Development of nanocomposites for bone grafting. Compos. Sci. Technol. 65, 2385 (2005).CrossRefGoogle Scholar
Macak, J.M., Tsuchiya, H., Taveira, L., Ghicov, A., and Schmuki, P.: Self-organized nanotubular oxide layers on Ti–6Al–7Nb and Ti–6Al–4V formed by anodization in NH4F solutions. J. Biomed. Mater. Res., Part A 75, 928 (2005).CrossRefGoogle ScholarPubMed
Gong, D., Grimes, C.A., Varghese, O.K., Hu, W., Singh, R.S., Chen, Z., and Dickey, E.C.: Titanium oxide nanotube arrays prepared by anodic oxidation. J. Mater. Res. 16, 3331 (2011).CrossRefGoogle Scholar
Kubies, D., Himmlová, L., Riedel, T., Chánová, E., Balík, K., Rová, M.D.Ě., Bártová, J., and Pešáková, V.: The interaction of osteoblasts with bone-implant materials: 1. The effect of physicochemical surface properties of implant materials. Physiol. Res. 8408, 95 (2011).CrossRefGoogle Scholar
Le Guehennec, L., Enkel, B., Weiss, P., Amouriq, Y., and Layrolle, P.: Osteoblastic cell behaviour on different titanium implant surfaces. Acta Biomater. 4, 535 (2008).CrossRefGoogle ScholarPubMed
Ehrenfest, D.M.D., Coelho, P.G., Kang, B., Sul, Y., and Albrektsson, T.: Classification of osseointegrated implant surfaces: Materials, chemistry and topography. Trends Biotechnol. 28, 198 (2010).CrossRefGoogle Scholar
Rieger, E., Dupret-bories, A., Salou, L., Layrolle, P., Debry, C., Lavalle, P., and Vrana, N.E.: Controlled implant/soft tissue interaction by nanoscale surface modifications of 3D porous titanium implants. Nanoscale 7, 9908 (2015).CrossRefGoogle ScholarPubMed
Wei, J., Igarashi, T., Okumori, N., Igarashi, T., Maetani, T., Liu, B., and Yoshinari, M.: Influence of surface wettability on competitive protein adsorption and initial attachment of osteoblasts. Biomed. Mater. 4, 45002 (2009).CrossRefGoogle ScholarPubMed
Hori, K. and Matsumoto, S.: Bacterial adhesion: From mechanism to control. Biochem. Eng. J. 48, 424 (2010).CrossRefGoogle Scholar
Neoh, K.G., Hu, X., Zheng, D., and Kang, E.T.: Balancing osteoblast functions and bacterial adhesion on functionalized titanium surfaces. Biomaterials 33, 2813 (2012).CrossRefGoogle ScholarPubMed
Yao, C. and Webster, T.J.: Prolonged antibiotic delivery from anodized nanotubular titanium using a co-precipitation drug loading method. J. Biomed. Mater. Res., Part B 91, 587 (2009).CrossRefGoogle ScholarPubMed
Unagolla, J.M. and Jayasuriya, A.C.: Drug transport mechanisms and in vitro release kinetics of vancomycin encapsulated chitosan–alginate polyelectrolyte microparticles as a controlled drug delivery system. Eur. J. Pharm. Sci. 114, 199 (2018).CrossRefGoogle ScholarPubMed
Wei, D., Zhou, R., Feng, W., Li, B., and Wang, Y.: Microarc oxidized TiO2 based ceramic coatings combined with cefazolin sodium/chitosan composited drug fi lm on porous titanium for biomedical applications. Mater. Sci. Eng., C 33, 4118 (2013).CrossRefGoogle Scholar
Mohan, L., Anandan, C., and Rajendran, N.: Drug release characteristics of quercetin-loaded TiO2 nanotubes coated with chitosan. Int. J. Biol. Macromol. 93, 1633 (2016).CrossRefGoogle ScholarPubMed
Shoulders, M.D. and Raines, R.T.: Collagen structure and stability. Annu. Rev. Biochem. 78, 929 (2009).CrossRefGoogle ScholarPubMed
Roehlecke, C., Witt, M., Kasper, M., Schulze, E., Wolf, C., Hofer, A., and Funk, R.H.W.: Synergistic effect of titanium alloy and collagen type I on cell adhesion, proliferation and differentiation of osteoblast-like cells. Cells Tissues Organs 168, 178 (2001).CrossRefGoogle ScholarPubMed
Landis, W.J. and Silver, F.H.: Mineral deposition in the extracellular matrices of vertebrate tissues: Identification of possible apatite nucleation sites on type I collagen. Cells Tissues Organs 189, 20 (2009).CrossRefGoogle ScholarPubMed
Macak, J.M., Tsuchiya, H., and Schmuki, P.: High-aspect-ratio TiO2 nanotubes by anodization of titanium Angew. Chem., Int. Ed. 44, 2100 (2005).Google Scholar
Macak, J.M., Tsuchiya, H., Ghicov, A., Yasuda, K., Hahn, R., Bauer, S., and Schmuki, P.: TiO2 nanotubes: Self-organized electrochemical formation, properties and applications. Curr. Opin. Solid State Mater. Sci. 11, 3 (2007).CrossRefGoogle Scholar
Regonini, D., Bowen, C.R., Jaroenworaluck, A., and Stevens, R.: A review of growth mechanism, structure and crystallinity of anodized TiO2 nanotubes. Mater. Sci. Eng., R 74, 377 (2013).CrossRefGoogle Scholar
Ghicov, A. and Schmuki, P.: Self-ordering electrochemistry: A review on growth and functionality of TiO2 nanotubes and other self-aligned MOx structures. Chem. Commun. 20, 2791 (2009).CrossRefGoogle Scholar
Mohammed, M.T., Khan, Z.A., and Siddiquee, A.N.: Surface modifications of titanium materials for developing corrosion behavior in human body environment: A review. Procedia Mater. Sci. 6, 1610 (2014).CrossRefGoogle Scholar
Kulkarni, M., Mazare, A., Schmuki, P., and Iglič, A.: Biomaterial surface modification of titanium and titanium alloys for medical applications. Nanomedicine 45, 111 (2017).Google Scholar
Scherrer, P.: Determination of the size and internal structure of colloidal particles using X-rays. Math-Phys Klasse 2, 98 (1918).Google Scholar
Shin, D.H., Shokuhfar, T., Choi, C.K., Lee, S.H., and Friedrich, C.: Wettability changes of TiO2 nanotube surfaces. Nanotechnology 22, 315704315711 (2011).CrossRefGoogle ScholarPubMed
Sun, T. and Wang, M.: A comparative study on titania layers formed on Ti, Ti–6Al–4V and NiTi shape memory alloy through a low temperature oxidation process. Surf. Coat. Technol. 205, 92 (2010).CrossRefGoogle Scholar
Ritger, P.L. and Peppas, N.A.: A simple equation for description of solute release II. Fickian and anomalous release from swellable devices 5, 23 (1987).CrossRefGoogle Scholar
Feng, W., Geng, Z., Li, Z., Cui, Z., Zhu, S., Liang, Y., Liu, Y., Wang, R., and Yang, X.: Controlled release behaviour and antibacterial effects of antibiotic-loaded titania nanotubes. Mater. Sci. Eng., C 62, 105 (2016).CrossRefGoogle ScholarPubMed
Zitting, A. and Skyttä, E.: Biological activity of titanium dioxides. Int. Arch. Occup. Environ. Health 43, 93 (1979).CrossRefGoogle ScholarPubMed
Simon, A.P. and Sikora, M.d.S.: Eletrólito para síntese eletroquímica de biomateriais a base de titânio. BR Patent No. 10 2018 002514 7, 2018.Google Scholar
Kokubo, T., Kim, H., and Kawashita, M.: Novel bioactive materials with different mechanical properties. Biomaterials 24, 2161 (2003).CrossRefGoogle ScholarPubMed
Schneider, C.A., Rasband, W.S., Eliceiri, K.W., and Instrumentation, C.: NIH image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671 (2012).CrossRefGoogle ScholarPubMed
Gulati, K., Kogawa, M., Prideaux, M., Findlay, D.M., Atkins, G.J., and Losic, D.: Drug-releasing nano-engineered titanium implants: Therapeutic efficacy in 3D cell culture model, controlled release and stability. Mater. Sci. Eng., C 69, 831 (2016).CrossRefGoogle ScholarPubMed
Abdou, H.M.: Dissolution, bioavailability and bioequivalence. Drug Dev. Ind. Pharm. 16, 185 (1990).CrossRefGoogle Scholar
Sinn Aw, M., Kurian, M., and Losic, D.: Non-eroding drug-releasing implants with ordered nanoporous and nanotubular structures: Concepts for controlling drug release. Biomater. Sci. 2, 10 (2014).CrossRefGoogle Scholar
Clinical and Laboratory Standards Institute: Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically (Clinical and Laboratory Standards Institute, Wayne, Pennsylvania, 2015).Google Scholar
Krichen, F., Karoud, W., Sila, A., Abdelmalek, B.E., Ghorbel, R., Ellouz-Chaabouni, S., and Bougatef, A.: Extraction, characterization and antimicrobial activity of sulfated polysaccharides from fish skins. Int. J. Biol. Macromol. 75, 283 (2015).CrossRefGoogle ScholarPubMed
Bulmus, V., Woodward, M., Lin, L., Murthy, N., Stayton, P., and Hoffman, A.: A new pH-responsive and glutathione-reactive, endosomal membrane-disruptive polymeric carrier for intracellular delivery of biomolecular drugs. J. Controlled Release 93, 105 (2003).CrossRefGoogle ScholarPubMed
Palanivelu, R. and Ruban Kumar, A.: Synthesis, characterization, in vitro anti-proliferative and hemolytic activity of hydroxyapatite. Spectrochim. Acta, Part A 127, 434 (2014).CrossRefGoogle ScholarPubMed